Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Grete F. Lauritzsen is active.

Publication


Featured researches published by Grete F. Lauritzsen.


Blood | 2008

Long-term progression-free survival of mantle cell lymphoma after intensive front-line immunochemotherapy with in vivo–purged stem cell rescue: a nonrandomized phase 2 multicenter study by the Nordic Lymphoma Group

Christian H. Geisler; Arne Kolstad; Anna Laurell; Niels S. Andersen; Lone Bredo Pedersen; Mats Jerkeman; Mikael Eriksson; Marie Nordström; Eva Kimby; Anne Marie Boesen; Outi Kuittinen; Grete F. Lauritzsen; Herman Nilsson-Ehle; Elisabeth Ralfkiaer; Måns Åkerman; Mats Ehinger; Christer Sundström; Ruth Langholm; Jan Delabie; Marja-Liisa Karjalainen-Lindsberg; Peter de Nully Brown; Erkki Elonen

Mantle cell lymphoma (MCL) is considered incurable. Intensive immunochemotherapy with stem cell support has not been tested in large, prospective series. In the 2nd Nordic MCL trial, we treated 160 consecutive, untreated patients younger than 66 years in a phase 2 protocol with dose-intensified induction immunochemotherapy with rituximab (R) + cyclophosphamide, vincristine, doxorubicin, prednisone (maxi-CHOP), alternating with R + high-dose cytarabine. Responders received high-dose chemotherapy with BEAM or BEAC (carmustine, etoposide, cytarabine, and melphalan/cyclophosphamide) with R-in vivo purged autologous stem cell support. Overall and complete response was achieved in 96% and 54%, respectively. The 6-year overall, event-free, and progression-free survival were 70%, 56%, and 66%, respectively, with no relapses occurring after 5 years. Multivariate analysis showed Ki-67 to be the sole independent predictor of event-free survival. The nonrelapse mortality was 5%. The majority of stem cell products and patients assessed with polymerase chain reaction (PCR) after transplantation were negative. Compared with our historical control, the Nordic MCL-1 trial, the event-free, overall, and progression-free survival, the duration of molecular remission, and the proportion of PCR-negative stem cell products were significantly increased (P < .001). Intensive immunochemotherapy with in vivo purged stem cell support can lead to long-term progression-free survival of MCL and perhaps cure. Registered at www.isrctn.org as #ISRCTN 87866680.


Journal of Clinical Oncology | 2012

Up-Front Autologous Stem-Cell Transplantation in Peripheral T-Cell Lymphoma: NLG-T-01

Francesco d'Amore; Thomas Relander; Grete F. Lauritzsen; Esa Jantunen; Hans Hagberg; Harald Anderson; Harald Holte; Anders Österborg; Mats Merup; Peter Brown; Outi Kuittinen; Martin Erlanson; Bjørn Østenstad; Unn-Merete Fagerli; Ole Gadeberg; Christer Sundström; Jan Delabie; Elisabeth Ralfkiaer; Martine Vornanen; Helle Toldbod

PURPOSE Systemic peripheral T-cell lymphomas (PTCLs) respond poorly to conventional therapy. To evaluate the efficacy of a dose-dense approach consolidated by up-front high-dose chemotherapy (HDT) and autologous stem-cell transplantation (ASCT) in PTCL, the Nordic Lymphoma Group (NLG) conducted a large prospective phase II study in untreated systemic PTCL. This is the final report, with a 5-year median follow-up, of the NLG-T-01 study. PATIENTS AND METHODS Treatment-naive patients with PTCL age 18 to 67 years (median, 57 years) were included. Anaplastic lymphoma kinase (ALK) -positive anaplastic large-cell lymphoma (ALCL) was excluded. An induction regimen of six cycles of biweekly CHOEP (cyclophosphamide, doxorubicin, vincristine, etoposide, and prednisone) was administered (in patients age > 60 years, etoposide was omitted). If in complete or partial remission, patients proceeded to consolidation with HDT/ASCT. RESULTS Of 166 enrolled patients, 160 had histopathologically confirmed PTCL. The majority presented with advanced-stage disease, B symptoms, and elevated serum lactate dehydrogenase. A total of 115 underwent HDT/ASCT, with 90 in complete remission at 3 months post-transplantation. Early failures occurred in 26%. Treatment-related mortality was 4%. At 60.5 months of median follow-up, 83 patients were alive. Consolidated 5-year overall and progression-free survival (PFS) were 51% (95% CI, 43% to 59%) and 44% (95% CI, 36% to 52%), respectively. Best results were obtained in ALK-negative ALCL. CONCLUSION Dose-dense induction followed by HDT/ASCT was well tolerated and led to long-term PFS in 44% of treatment-naive patients with PTCL. This represents an encouraging outcome, particularly considering the high median age and adverse risk profile of the study population. Therefore, dose-dense induction and HDT/ASCT are a rational up-front strategy in transplantation-eligible patients with PTCL.


British Journal of Haematology | 2012

Nordic MCL2 trial update: six-year follow-up after intensive immunochemotherapy for untreated mantle cell lymphoma followed by BEAM or BEAC + autologous stem-cell support: still very long survival but late relapses do occur

Christian H. Geisler; Arne Kolstad; Anna Laurell; Mats Jerkeman; Riikka Räty; Niels S. Andersen; Lone Bredo Pedersen; Mikael Eriksson; Marie Nordström; Eva Kimby; Hans Bentzen; Outi Kuittinen; Grete F. Lauritzsen; Herman Nilsson-Ehle; Elisabeth Ralfkiaer; Mats Ehinger; Christer Sundström; Jan Delabie; Marja-Liisa Karjalainen-Lindsberg; Peter de Nully Brown; Erkki Elonen

Mantle cell lymphoma (MCL) is a heterogenic non‐Hodgkin lymphoma entity, with a median survival of about 5 years. In 2008 we reported the early – based on the median observation time of 4 years – results of the Nordic Lymphoma Group MCL2 study of frontline intensive induction immunochemotherapy and autologous stem cell transplantation (ASCT), with more than 60% event‐free survival at 5 years, and no subsequent relapses reported. Here we present an update after a median observation time of 6·5 years. The overall results are still excellent, with median overall survival and response duration longer than 10 years, and a median event‐free survival of 7·4 years. However, six patients have now progressed later than 5 years after end of treatment. The international MCL Prognostic Index (MIPI) and Ki‐67‐expression were the only independent prognostic factors. Subdivided by the MIPI‐Biological Index (MIPI + Ki‐67, MIPI‐B), more than 70% of patients with low‐intermediate MIPI‐B were alive at 10 years, but only 23% of the patients with high MIPI‐B. These results, although highly encouraging regarding the majority of the patients, underline the need of a risk‐adapted treatment strategy for MCL. The study was registered at www.isrctn.org as ISRCTN 87866680.


Blood | 2010

The Mantle Cell Lymphoma International Prognostic Index (MIPI) is superior to the International Prognostic Index (IPI) in predicting survival following intensive first-line immunochemotherapy and autologous stem cell transplantation (ASCT)

Christian H. Geisler; Arne Kolstad; Anna Laurell; Riikka Räty; Mats Jerkeman; Mikael Eriksson; Marie Nordström; Eva Kimby; Anne Marie Boesen; Herman Nilsson-Ehle; Outi Kuittinen; Grete F. Lauritzsen; Elisabeth Ralfkiaer; Mats Ehinger; Christer Sundström; Jan Delabie; Marja-Liisa Karjalainen-Lindsberg; Peter Brown; Erkki Elonen

Mantle cell lymphoma (MCL) has a heterogeneous clinical course. The recently proposed Mantle Cell Lymphoma International Prognostic Index (MIPI) predicted the survival of MCL better than the International Prognostic Index in MCL patients treated with conventional chemotherapy, but its validity in MCL treated with more intensive immunochemotherapy has been questioned. Applied here to 158 patients of the Nordic MCL2 trial of first-line intensive immunochemotherapy followed by high-dose chemotherapy and autologous stem cell transplantation, the MIPI and the simplified MIPI (s-MIPI) predicted survival significantly better (P < .001) than the International Prognostic Index (P > .004). Both the MIPI and the s-MIPI mainly identified 2 risk groups, low and intermediate versus high risk, with the more easily applied s-MIPI being just as powerful as the MIPI. The MIPI(B) (biological), incorporating Ki-67 expression, identified almost half of the patients as high risk. We suggest that also a simplified MIPI(B) is feasible.


Journal of Clinical Oncology | 2009

Pre-Emptive Treatment With Rituximab of Molecular Relapse After Autologous Stem Cell Transplantation in Mantle Cell Lymphoma

Niels S. Andersen; Lone Bredo Pedersen; Anna Laurell; Erkki Elonen; Arne Kolstad; Anne Marie Boesen; Lars Møller Pedersen; Grete F. Lauritzsen; Roald Ekanger; Herman Nilsson-Ehle; Marie Nordström; Susanne Fredén; Mats Jerkeman; Mikael Eriksson; Jaan Väärt; Beatrice Malmer; Christian H. Geisler

PURPOSE Minimal residual disease (MRD) is predictive of clinical progression in mantle-cell lymphoma (MCL). According to the Nordic MCL-2 protocol we prospectively analyzed the efficacy of pre-emptive treatment using rituximab to MCL patients in molecular relapse after autologous stem cell transplantation (ASCT). PATIENTS AND MATERIALS MCL patients enrolled onto the study, who had polymerase chain reaction (PCR) detectable molecular markers and underwent ASCT, were followed with serial PCR assessments of MRD in consecutive bone marrow and peripheral blood samples after ASCT. In case of molecular relapse with increasing MRD levels, patients were offered pre-emptive treatment with rituximab 375 mg/m(2) weekly for 4 weeks. RESULTS Of 160 MCL patients enrolled, 145 underwent ASCT, of whom 78 had a molecular marker. Of these, 74 were in complete remission (CR) and four had progressive disease after ASCT. Of the CR patients, 36 underwent a molecular relapse up to 6 years (mean, 18.5 months) after ASCT. Ten patients did not receive pre-emptive treatment mainly due to a simultaneous molecular and clinical relapse, while 26 patients underwent pre-emptive treatment leading to reinduction of molecular remission in 92%. Median molecular and clinical relapse-free survival after pre-emptive treatment were 1.5 and 3.7 years, respectively. Of the 38 patients who remain in molecular remission for now for a median of 3.3 years (range, 0.4 to 6.6 years), 33 are still in clinical CR. CONCLUSION Molecular relapse may occur many years after ASCT in MCL, and PCR based pre-emptive treatment using rituximab is feasible, reinduce molecular remission, and may prevent clinical relapse.


International Journal of Cancer | 1998

Clonal deletion of thymocytes as a tumor escape mechanism

Grete F. Lauritzsen; Peter O. Hofgaard; Karl Schenck; Bjarne Bogen

Clonal deletion of thymocytes is a major event in T‐cell tolerance and might represent a tumor escape mechanism. Previously, we have shown that class II‐restricted, Id‐specific, CD4+ T cells in T‐cell receptor (TCR)‐transgenic mice confer resistance against the MOPC315 plasmacytoma. In this report, we have investigated whether monoclonal immunoglobulin (Ig) produced by a plasmacytoma can induce deletion of thymocytes specific for the variable parts of Ig, i.e., the idiotype (Id). Large numbers of MOPC315 tumor cells were injected s.c. in the TCR‐transgenic mice to overwhelm the CD4+ T‐cell‐mediated protection. When the MOPC315 plasmacytomas reached a weight of approximately 0.5 g (serum myeloma protein M315 about 50 μg/ml), immature CD4+8+ and mature CD4+ transgenic thymocytes became progressively deleted. Apoptotic thymocytes were already detectable when tumors were 2 mm in diameter (serum M315: 5 μg/ml, or 0.03 μM). The negative selection was Id‐specific, because an Id‐negative plasmacytoma failed to induce deletion. Injection of purified MOPC315‐myeloma protein (M315) i.p. caused a profound reduction of Id‐specific thymocytes. Enriched thymic dendritic cells (DC) from tumor‐bearing animals were found to be primed with λ2315 and induced apoptosis of thymocytes in vitro. Our results indicate that circulating myeloma protein is processed and presented by thymic antigen‐presenting cells (APC), and induces deletion of Id‐specific thymocytes. Deletion of tumor‐specific thymocytes may represent a tumor escape mechanism in patients with cancers that secrete or shed tumor antigens. The possibility that vaccination with tumor Ig or genes encoding for it may induce tolerance instead of protection should be taken into consideration. Int. J. Cancer 78:216–222, 1998.© 1998 Wiley‐Liss, Inc.


Blood | 2014

Nordic MCL3 study: 90Y-ibritumomab-tiuxetan added to BEAM/C in non-CR patients before transplant in mantle cell lymphoma

Arne Kolstad; Anna Laurell; Mats Jerkeman; Kirsten Grønbæk; Erkki Elonen; Riikka Räty; Lone Bredo Pedersen; Annika Loft; Trond Velde Bogsrud; Eva Kimby; Per Boye Hansen; Unn-Merete Fagerli; Herman Nilsson-Ehle; Grete F. Lauritzsen; A. K. Lehmann; Christer Sundström; Marja-Liisa Karjalainen-Lindsberg; Elisabeth Ralfkiaer; Mats Ehinger; Jan Delabie; Hans Bentzen; Jukka Schildt; Kamelia Kostova-Aherdan; Henrik Frederiksen; Peter Brown; Christian H. Geisler

The main objective of the MCL3 study was to improve outcome for patients not in complete remission (CR) before transplant by adding (90)Y-ibritumomab-tiuxetan (Zevalin) to the high-dose regimen. One hundred sixty untreated, stage II-IV mantle cell lymphoma patients <66 years received rituximab (R)-maxi-CHOP (cyclophosphamide, hydroxydaunorubicin, vincristine, and prednisone) alternating with R-high-dose cytarabine (6 cycles total), followed by high-dose BEAM/C (bis-chloroethylnitrosourea, etoposide, cytarabine, and melphalan or cyclophosphamide) and autologous stem cell transplantation from 2005 to 2009. Zevalin (0.4 mCi/kg) was given to responders not in CR before transplant. Overall response rate pretransplant was 97%. The outcome did not differ from that of the historic control: the MCL2 trial with similar treatment except for Zevalin. Overall survival (OS), event-free survival (EFS), and progression-free survival (PFS) at 4 years were 78%, 62%, and 71%, respectively. For responding non-CR patients who received Zevalin, duration of response was shorter than for the CR group. Inferior PFS, EFS, and OS were predicted by positron emission tomography (PET) positivity pretransplant and detectable minimal residual disease (MRD) after transplant. In conclusion, positive PET and MRD were strong predictors of outcome. Intensification with Zevalin may be too late to improve the outcome of patients not in CR before transplant. This trial was registered at www.clinicaltrials.gov as #NCT00514475.


British Journal of Haematology | 2011

High dose chemotherapy with autologous stem cell support for patients with histologically transformed B-cell non-Hodgkin lymphomas. A Norwegian multi centre phase II study

Marianne B. Eide; Grete F. Lauritzsen; Gunnar Kvalheim; Arne Kolstad; Unn M. Fagerli; Martin Maisenhölder; Bjørn Østenstad; Øystein Fluge; Jan Delabie; Harald Aarset; Knut Liestøl; Harald Holte

We present a prospective phase II study of patients with relapse after chemotherapy showing transformation of follicular lymphoma to diffuse large B‐cell lymphoma, performed before rituximab was included in standard treatment. Patients in complete (CR) or partial remission (PR) after salvage chemotherapy were eligible for high‐dose chemotherapy with autologous stem cell support (HDT). Forty‐seven patients from five Norwegian centres were included, of whom 30 (63%) received HDT. Eighteen (60%) achieved CR, seven (23%) PR and five (10%) had progressive disease following HDT. Median follow‐up for the surviving patients was 75 months; median progression‐free (PFS) and overall survival (OS) were 26 and 47 months, respectively. Median OS for all patients was 43 months, compared to only 10 months for patients not eligible for HDT. Patients receiving CD34+ enriched/B‐cell depleted grafts had inferior PFS and a trend for inferior OS compared to patients receiving non‐purged grafts (Log Rank 0·025 and 0·151, respectively). In conclusion, two thirds of patients with transformation of follicular lymphoma were eligible for HDT. The majority of patients achieved CR and a considerable number had prolonged OS. The use of in vitro purged grafts did not result in a survival benefit compared to that of non‐purged grafts.


The Journal of Molecular Diagnostics | 2004

Constitutive Expression of the AP-1 Transcription Factors c-jun, junD, junB, and c-fos and the Marginal Zone B-Cell Transcription Factor Notch2 in Splenic Marginal Zone Lymphoma

Gunhild Trøen; Vigdis Nygaard; Tor Kristian Jenssen; Ida Münster Ikonomou; Anne Tierens; Estella Matutes; Alicja M. Gruszka-Westwood; Daniel Catovsky; Ola Myklebost; Grete F. Lauritzsen; Eivind Hovig; Jan Delabie

Splenic marginal zone lymphoma (SMZL) is a lymphoma type of putative marginal zone B-cell origin. No specific genetic alterations have yet been demonstrated in SMZL. Clinically, SMZL is a low-grade B-cell non-Hodgkin lymphoma. However, the presence of p53 mutation, 7q22-7q32 deletion or the absence of somatic hypermutations of immunoglobulin genes has been correlated with a worse prognosis. In this study, we analyzed genome-wide gene expression of 24 cases of SMZL using the microarray technique. The AP-1 transcription factors c-jun, junD, junB, and c-fos as well as Notch2 were found to be specifically up-regulated. These data were confirmed by real-time PCR and immunohistochemical staining of tissue sections. The absence of concordant high expression of the MAP kinases, the signaling cascade leading to AP-1 up-regulation, suggests autoregulation of the AP-1 transcription factors and an important role in SMZL oncogenesis. High expression of Notch2, a transcription factor that induces marginal zone B-cell differentiation, is highly suggestive for a marginal zone B-cell origin of SMZL. In addition, SMZL with the 7q deletion showed high expression of TGF-beta1 and low expression of the DNA helicase XPB, a crucial part of the nucleotide excision repair complex, possibly explaining the more aggressive clinical course of those cases.


Leukemia & Lymphoma | 2007

Standard CHOP-21 as first line therapy for elderly patients with Hodgkin's lymphoma

Arne Kolstad; Ole Nome; Jan Delabie; Grete F. Lauritzsen; Alexander Fosså; Harald Holte

There is no consensus on the optimal chemotherapy regimen for Hodgkins lymphoma patients ≥ 60 years. We present our institutions results of 5 years, using CHOP-21 as standard for this patient group. Twenty-nine patients with a median age of 71 years (range, 60 – 91) were included in this cohort. Fifty-five percent had known co-morbidities. Stage I/IIA patients (38%) were treated with 2 – 4 cycles of CHOP followed by radiotherapy. Stage IIB – IV patients (62%) received 6 – 8 cycles of CHOP and for the majority (13/18 pts) no radiotherapy. Two treatment-related deaths occurred. Febrile neutropenia was the most common toxicity (31%). The complete response rate after CHOP +/− radiotherapy was 93%. With a median follow-up of 41 months, five patients have relapsed and four have died from Hodgkins lymphoma. So far, no relapses have occurred after 2 years from the end of therapy. Overall survival and progression-free survival at 3 years were 79% and 76%, respectively. We conclude that CHOP-21 is a well-tolerated and effective treatment for elderly patients with Hodgkins lymphoma.

Collaboration


Dive into the Grete F. Lauritzsen's collaboration.

Top Co-Authors

Avatar

Arne Kolstad

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harald Holte

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar

Unn-Merete Fagerli

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge